Angiogenesis is required for tumor growth and metastasis. It has recently been suggested that thrombin is a potent promoter of angiogenesis. We therefore examined the possibility that thrombin could be inducing the expression of angiopoietin-2 (Ang-2), necessary for remodeling. Human umbilical vein endothelial cells were incubated with or without thrombin (1 U/mL) for 1 to 24 hours and then examined for messenger RNA (mRNA) by Northern analysis. Enhanced mRNA expression (about 4-fold over baseline) was noted at 4 hours. Enhanced expression of Ang-2 mRNA was secondary to enhanced transcription (about 4-fold), with no effect on stabilization. Enhanced Ang-2 mRNA transcription was inhibited by H7 and PD98059, indicating the requirement of serine/threonine kinases as well as the mitogen-activated protein kinase pathway. Up-regulation of mRNA was associated with enhanced Ang-2 protein synthesis and secretion as assayed by immunoblot. Thrombin-induced secreted Ang-2 inhibited the binding of recombinant 35S–Ang-1 to its Tie-2–Fc receptor, demonstrating functionality. Hirudin reversed this effect, demonstrating thrombin specificity. Thus, thrombin-induced tumorigenesis and metastasis is associated with enhanced Ang-2 protein synthesis and secretion via enhanced transcription of Ang-2. This could help explain how thrombin promotes angiogenesis.

Angiogenesis, the process of new blood vessel growth, is essential for embryonic development as well as the support of normal metabolic demands. It is also essential in pathologic conditions, such as wound healing and tumor growth.1,2Various growth factors, particularly vascular endothelial growth factor (VEGF), angiopoietin-1 (Ang-1), and angiopoietin-2 (Ang-2), have been implicated in the regulation of vessel formation. VEGF promotes angiogenesis in vitro by inducing confluent microvascular endothelial cells to invade collagen gels and form capillarylike structures.3 VEGF consists of a family of 6 ligands (placental growth factor, VEGF, and VEGF B, C, D, and E) that bind to 3 receptor tyrosine kinases on endothelial cells (VEGFR-1/Flt-1, VEGFR-2/Flk-1/KDR, and VEGFR-3/Flt-4).4,5 

A second family of proteins exerting an important role in vascular development and stabilization has now been identified and designated angiopoietins. Angiopoietins regulate angiogenesis by activating or blocking the activation of Tie-2/Tek, a surface receptor tyrosine kinase generally restricted to endothelial cells. Ang-1 helps to maintain vascular integrity and is involved in vessel maturation.6-8 Ang-2 acts as an antagonist of the Ang-1/Tie2 interaction.9 Ang-2 is expressed in areas undergoing vascular remodeling and is involved in neovascularization. By competing with Ang-1 for binding to their common receptor Tie2, Ang-2 leads to decreased vessel maturation and either vessel regression (in the absence of VEGF) or enhanced vessel sprouting (in the presence of VEGF).10 Both hypoxia and VEGF up-regulate Ang-2 expression.11 

It has recently been suggested that thrombin is a potent promoter of angiogenesis.12-15 The relationship between thrombosis and cancer/metastasis was first recognized by Trousseau in 1872.16 Many studies have described a systemic activation of the blood coagulation cascade in patients with cancer.17-20 Recent studies have shown that thrombin-treated tumor cells have an increased ability to adhere to von Willebrand factor, fibronectin, platelets, and endothelial cells and form pulmonary metastases in syngeneic mice.21-25 However, the data do not define the mechanism by which thrombin promotes tumor progression and metastasis. Thrombin has a multitude of enzymatic and cellular actions on a variety of cell types.26 In this communication, we report the induction of Ang-2 messenger RNA (mRNA) and the synthesis and secretion of protein by thrombin and investigate its mechanism.

Cell culture and materials

Human diploid primary culture fibroblasts (FS4) were kindly provided by Dr J. Vilcek (New York University Medical Center, New York, NY) and cultured in Dulbecco modified Eagle medium with 10% fetal calf serum and 1% penicillin and streptomycin. Human umbilical vein endothelial (HUVE) cells were derived from fresh umbilical cord. The cord was washed 3 times with phosphate-buffered saline, treated with 0.5% trypsin (Sigma, St Louis, MO) for 3 to 5 minutes, and the HUVE cells isolated by washing and centrifugation in phosphate-buffered saline. HUVE cells were cultured in EBM-2 media (Clonetics) containing 10% fetal calf serum, 1% penicillin/streptomycin, and endothelial growth supplements (Sigma). Cells were starved for 4 hours in Dulbecco modified Eagle medium and then treated with different concentrations of thrombin for various time intervals. Thrombin, wortmannin, H7, and PD998059 were obtained from Sigma.

Northern Blot analysis

Total RNA was extracted, fractionated by electrophoresis on a 1% agarose gel in 6.7% formaldehyde, and transferred onto a Genescreen Plus nylon membrane (NEN Life Science Products, Boston, MA). The membrane was hybridized at 42°C overnight with32P-dCTP–labeled probes specific for Ang-2 (1.5 kb reverse transcriptase–polymerase chain reaction product, which contains the complete coding region). The blots were sequentially washed with varying dilutions of SSC, the last being 0.1× SSC at 65°C for 30 minutes. Autoradiography was carried out at −70°C with an intensifying screen. The autoradiographic signals were quantified by densitometric analysis (Personal Densitometer, Molecular Dynamics/Amersham).

Immunoblotting of secreted Ang-2

Cells were treated with 1 U/mL thrombin for 8 hours. Culture media were collected and concentrated with Centricom (Sigma), and cells were extracted in lysis buffer: 1% Triton X-100, 150 mM NaCl, 10 mM Tris (pH 7.4), 50 μg/mL pepstatin, 20 μg/mL aprotinin, 2 μg/mL leupeptin, 0.2 mM phenylmethylsulfonyl fluoride, 1 mM ethylenediaminetetraacetic acid, 0.2 mM sodium orthovanadate, and 0.5% Nonidet P-40. A total of 100 μg of protein, determined by BioRad protein assay, was run on sodium dodecyl sulfate (SDS)–polyacrylamide gel, transferred to a membrane, incubated with blocking solution (2% powdered milk, 0.2% Tween 20 in phosphate-buffered saline), and reacted with a specific goat anti–Ang-2 polyclonal antibody (Santa Cruz, CA). After washing, peroxidase-conjugated second antibodies were applied and chemiluminescence generated by incubation with ECL reagents (Amersham Life Science).

Nuclear run-on analysis of transcription

HUVE cells were stimulated with or without thrombin (1 U/mL) for 1 hour. The cells were then lysed and nuclei isolated by centrifugation at 12 000g for 15 minutes at 4°C. The nuclear suspension was incubated with 0.5 mM each of CTP, ATP, and GTP and with 250 μCi (9.25 MBq) of 32P-UTP. The samples were extracted with phenol/chloroform, precipitated, and resuspended at equal counts per milliliter in hybridization buffer. Denatured plasmid DNA harboring the Ang-2 fragment as well as GAPDH were blotted onto nitrocellulose filters and then hybridized with the radiolabeled samples. The filters were washed as described above for Northern analysis and the membranes subjected to autoradiography.

Actinomycin D chase studies

Confluent cells were incubated in the presence of thrombin (1 U/mL) for 1 hour at 37°C. The cells were then treated with media containing actinomycin D (5 μg/mL) and incubated for an additional 1 to 7 hours. Total RNA was extracted and analyzed for Ang-2 mRNA levels by Northern blot analysis.

Ang-2 inhibition of binding of Ang-1 to Tie-2–Fc

35S-labeled recombinant Ang-1 was prepared by in vitro translation of the complementary DNA27 using the coupled TNT transcription/translation ribosomal system (Promega, Madison, WI). The recombinant Tie-2–Fc fusion protein was prepared from 293 cells and purified with protein A beads as previously described.6 Cultured HUVE cells were treated with thrombin (1 U/mL) or thrombin plus hirudin (1:1) for 8 hours at 37°C. The media were removed and concentrated 10-fold with Centricon (AMICON, Beverly, MA). A total of 1 mL concentrated media was mixed with 5 μL recombinant 35S–Ang-1, incubated with 0.25 μg Tie-2–Fc for 3 hours at 4°C, and then precipitated with protein A beads and run on 12% SDS–polyacrylamide gel electrophoresis for autoradiography as described.27 

Effect of thrombin on the expression of Ang-2 mRNA

Because thrombin stimulates tumor growth and metastasis and enhanced angiogenesis is required for both, we investigated the effect of thrombin on the induction of Ang-2 mRNA, an angiogenesis growth factor required for blood vessel remodeling. Figure1 demonstrates that the expression of Ang-2 mRNA was upregulated about 4-fold by thrombin in HUVE cells with peak at 4 hours. Thrombin specificity was demonstrated by the more than 3-fold inhibition of Ang-2 mRNA with hirudin in HUVE cells (Figure 2).

Fig. 1.

Thrombin-induced increased expression of Ang-2 mRNA in HUVE cells.

Cells were starved in media containing 1% fetal calf serum overnight and then treated with or without thrombin for various time intervals. Cells were collected and analyzed for Ang-2 mRNA by Northern blot. Ribosomal 28S RNA is shown as an internal gel-loading standard.

Fig. 1.

Thrombin-induced increased expression of Ang-2 mRNA in HUVE cells.

Cells were starved in media containing 1% fetal calf serum overnight and then treated with or without thrombin for various time intervals. Cells were collected and analyzed for Ang-2 mRNA by Northern blot. Ribosomal 28S RNA is shown as an internal gel-loading standard.

Close modal
Fig. 2.

Thrombin specificity of Ang-2 mRNA up-regulation.

Cells were treated without (C) or with 1 U/mL thrombin (T) or with thrombin plus hirudin (T+H) for 3 hours and then analyzed by Northern blot.

Fig. 2.

Thrombin specificity of Ang-2 mRNA up-regulation.

Cells were treated without (C) or with 1 U/mL thrombin (T) or with thrombin plus hirudin (T+H) for 3 hours and then analyzed by Northern blot.

Close modal

Thrombin increases the production and secretion of Ang-2 protein

To define whether the up-regulation of Ang-2 mRNA was accompanied by an increase in protein synthesis, cells were cultured in media without or with thrombin (1 U/mL) for 8 hours. The cells were extracted with lysis buffer and the media collected and concentrated. Both were analyzed for Ang-2 production by Western blot. The production (Figure 3A) and secretion (Figure 3B) of Ang-2 was increased 2.5- and 3.5-fold, respectively, in HUVE cells.

Fig. 3.

Effect of thrombin on Ang-2 protein production and secretion.

FS4 and HUVE cells were cultured in media with or without thrombin, 1 U/mL, for 8 hours at 37°C. The media were collected and concentrated and the cells extracted with lysis buffer. Both were then analyzed for Ang-2 production by Western blot. (A) Ang-2 in HUVE cells, not in FS4 cells. (B) Ang-2 in supernatant of HUVE cells.

Fig. 3.

Effect of thrombin on Ang-2 protein production and secretion.

FS4 and HUVE cells were cultured in media with or without thrombin, 1 U/mL, for 8 hours at 37°C. The media were collected and concentrated and the cells extracted with lysis buffer. Both were then analyzed for Ang-2 production by Western blot. (A) Ang-2 in HUVE cells, not in FS4 cells. (B) Ang-2 in supernatant of HUVE cells.

Close modal

Thrombin induces increased transcription of Ang-2 mRNA

Thrombin-induced increased expression of Ang-2 mRNA could reflect increased gene transcription, increased mRNA stability, or both. To determine whether the increased Ang-2 mRNA was a result of increased gene transcription, cells were examined by nuclear run-on experiments. As shown in Figure 4, the rate of transcription of Ang-2 was increased by 3-fold in HUVE cells stimulated with thrombin.

Fig. 4.

Effect of thrombin on nuclear transcription of Ang-2 mRNA.

HUVE cells were treated with and without 1 U/mL thrombin for 3 hours, lysed, and nuclei extracted. Radiolabelled run-on RNAs were synthesized from isolated nuclei and hybridized to immobilized complementary DNA for Ang-2 and GAPDH (internal loading control).

Fig. 4.

Effect of thrombin on nuclear transcription of Ang-2 mRNA.

HUVE cells were treated with and without 1 U/mL thrombin for 3 hours, lysed, and nuclei extracted. Radiolabelled run-on RNAs were synthesized from isolated nuclei and hybridized to immobilized complementary DNA for Ang-2 and GAPDH (internal loading control).

Close modal

As shown in Figure 5, the mRNA stability of Ang-2 was not increased with thrombin in HUVE cells, although the level of expression of Ang-2 was clearly enhanced upon thrombin stimulation.

Fig. 5.

Effect of thrombin on Ang-2 mRNA stability.

Cells were treated with and without 1 U/mL thrombin for 3 hours, followed by addition of 5 μg/mL actinomycin D to inhibit RNA synthesis. Northern analysis was then performed at various time intervals of 1 to 24 hrs.

Fig. 5.

Effect of thrombin on Ang-2 mRNA stability.

Cells were treated with and without 1 U/mL thrombin for 3 hours, followed by addition of 5 μg/mL actinomycin D to inhibit RNA synthesis. Northern analysis was then performed at various time intervals of 1 to 24 hrs.

Close modal

Thrombin promotes the expression of Ang-2 via the serine/threonine kinase and MAPK pathways

To define the signaling pathways responsible for the increased expression of Ang-2 by thrombin, 3 major cellular transduction mechanisms involved in thrombin receptor activation28-30were studied (phosphatidylinositol-3 [PI-3] kinase, serine/threonine kinases, and mitogen-activated protein kinase [MAPK]). As shown in Figure 6, the increased expression of Ang-2 by thrombin in HUVE cells is regulated by 2 of the 3 pathways examined. Ang-2 induction was totally inhibited by H7 (serine/threonine kinase inhibitor) and by PD98059 (MAPK kinase inhibitor) (3-fold less for both) but not inhibited by wortmannin (PI-3 kinase inhibitor). These data indicate that the mechanism of thrombin-induced up-regulation of Ang-2 in HUVE cells is associated with activation of serine/threonine kinases and the MAPK pathway.

Fig. 6.

Effect of signaling pathway protein kinase inhibitors on thrombin-induced expression of Ang-2 mRNA.

Cells were treated without (C) or with wortmannin (W) 1 μM, H7 (20 μM), or PD98059 (100 μM) for 2.5 hours at 37°C and thrombin, 1 U/mL, added for an additional 3 hours. Northern blot was then performed for Ang-2 mRNA. Note inhibition by H7 and PD98059, which inhibit serine/threonine kinases and the MAPK pathway.

Fig. 6.

Effect of signaling pathway protein kinase inhibitors on thrombin-induced expression of Ang-2 mRNA.

Cells were treated without (C) or with wortmannin (W) 1 μM, H7 (20 μM), or PD98059 (100 μM) for 2.5 hours at 37°C and thrombin, 1 U/mL, added for an additional 3 hours. Northern blot was then performed for Ang-2 mRNA. Note inhibition by H7 and PD98059, which inhibit serine/threonine kinases and the MAPK pathway.

Close modal

Thrombin-induced Ang-2 inhibits the binding of Ang-1 to its receptor, Tie-2. Functional evidence that the thrombin-induced Ang-2 secretion is biologically active is provided by the following experiment. Recombinant 35S-labeled Ang-1 produced from an in vitro transcription/translation ribosomal system was shown to bind to its recombinant fusion protein receptor Tie-2–Fc (Figure7, lane 1). Ang-2 derived from the supernatants of thrombin-treated HUVE cells was then preincubated with35S–Ang-1 to incubation with Tie-2–Fc by Ang-2. Figure 7, lane 3, demonstrates inhibition of binding of Ang-1 to Tie-2–Fc by Ang-2. Figure 7, lane 2, demonstrates reversibility of the thrombin effect with hirudin.

Fig. 7.

Effect of thrombin-induced Ang-2 on binding of Ang-1 to its Tie-2 receptor.

HUVE cells were treated with 1 U/mL thrombin for 5 hours and the concentrated supernatant incubated with 35S–Ang-1 prior to binding of Ang-1 to Tie-2–Fc. Binding was analyzed by autoradiography following immunoprecipitation with protein A and 12% SDS–polyacrylamide gel electrophoresis. Lane 1, 35S–Ang-1 plus Tie-2–Fc. Lane 2, 35S–Ang-1 plus supernatant of thrombin-hirudin–treated HUVE cells. Lane 3, 35S–Ang-1 plus supernatant of thrombin-treated HUVE cells. Lane 4, absence of Tie-2–Fc in the system. Representative of 3 experiments.

Fig. 7.

Effect of thrombin-induced Ang-2 on binding of Ang-1 to its Tie-2 receptor.

HUVE cells were treated with 1 U/mL thrombin for 5 hours and the concentrated supernatant incubated with 35S–Ang-1 prior to binding of Ang-1 to Tie-2–Fc. Binding was analyzed by autoradiography following immunoprecipitation with protein A and 12% SDS–polyacrylamide gel electrophoresis. Lane 1, 35S–Ang-1 plus Tie-2–Fc. Lane 2, 35S–Ang-1 plus supernatant of thrombin-hirudin–treated HUVE cells. Lane 3, 35S–Ang-1 plus supernatant of thrombin-treated HUVE cells. Lane 4, absence of Tie-2–Fc in the system. Representative of 3 experiments.

Close modal

The requirement of angiogenesis for tumor growth and metastasis is well documented.1 The effect of thrombin and activated platelets on the promotion of tumor growth and metastasis is similarly well documented.21-25 The role of thrombin in the induction of angiogenesis has recently been explored by several groups and can now be attributed to its effects on VEGF, Ang-1, and Ang-2.

Platelets contain VEGF31,32 and Ang-1,27,33which are released following platelet activation with thrombin. Tumor specimens are surrounded by platelets and when removed at surgery have VEGF and thrombin localized on their surface.34-37 Tumor growth is inhibited by VEGF antibody.38,39 The recombinant Tie-2 receptor, AdExTek, capable of blocking Tie-2 activation by Ang-1, inhibits the growth and metastasis of murine mammary carcinoma (4T1) and melanoma (B16F10.9) cells.40 Ang-2 has been found in hypervascular human hepatocarcinomas as well as in an animal model in which it was highly expressed only in tumor tissue. Ectopic expression of Ang-2 in nonexpressing human hepatocellular cells promoted hepatomas in nude mice.41 Ang-2 has been found in advanced-stage neuroblastoma compared with low-stage tumors as well as in neuroblastoma cell lines,42 in increased intensity in blood vessels of non–small cell lung carcinoma,43uveal melanoma cell lines,44 thyroid tumor progression,45 and endothelial cells of human gliomas.46 

However, these associations do not define the mechanism by which thrombin promotes angiogenesis. Recent publications contribute to our understanding of this mechanism. Maragoudakis and coworkers have reported that thrombin promotes endothelial cell alignment in vitro in matrigel and angiogenesis in vivo,12 that angiogenesis is independent of fibin formation,15 and that thrombin potentiates VEGF by upregulating its recepor, KDR.14Herbert et al have demonstrated up-regulation of endothelial cell growth by thrombin by the autocrine release of basic fibroblast growth factor.13 Our group47,48 and Ollivier et al49 have recently demonstrated up-regulation of VEGF mRNA and protein in prostate DU145 cells47 and fibroblasts.47,49 We have recently demonstrated the presence of Ang-1 in platelets27 and its release by thrombin.33 

The role of Ang-2 in tumorigenesis and metastasis has recently been more clearly defined by Yancopoulos and coworkers.50 They have observed that tumor cells do not initially require vascular support but then proceed to co-opt existing host endothelial cells in which Ang-2 is highly induced prior to VEGF induction. The co-opted vessels then regress via disruption of endothelial cell interactions and undergo apoptosis, resulting in central necrosis of the tumor. Angiogenesis is then induced at the tumor margin associated with the induction of VEGF and Ang-2, supporting further growth.

Our study indicates, for the first time, up-regulation of functional Ang-2 by thrombin. Our current report on the induction of Ang-2 protein synthesis and secretion contributes to an understanding of the mechanism of thrombin-induced angiogenesis, tumor growth, and metastasis. Unlike the effect of thrombin on up-regulation of VEGF via enhanced stabilization of VEGF mRNA,47 Ang-2 upregulates via enhanced transcription, with absence of enhanced stabilization of mRNA. It has recently been reported by Oh et al11 that both hypoxia and VEGF up-regulate Ang-2 mRNA in bovine microvascular endothelial cells and that VEGF is capable of upregulating Ang-2. One should therefore consider the possibility that thrombin-induced up-regulation of Ang-2 may be secondary to its up-regulation of VEGF. However, this was shown not to be the case by these authors,24 who demonstrated that neutralizing anti-VEGF antibody had no effect on anoxia-induced up-regulation of Ang-2. Our studies similarly do not support this possibility because up-regulation of VEGF mRNA by thrombin is inhibited by wortmannin (PI-3 kinase inhibitor),47 whereas up-regulation of Ang-2 mRNA is not and HUVE cells have no detectable VEGF mRNA before or after thrombin stimulation.

Up-regulation of Ang-2 mRNA was inhibited by both serine/threonine kinase and a highly specific MAPK kinase inhibitor. Both signaling pathways are involved in thrombin stimulation of cells. Thrombin-induced cell protection of astrocytes is inhibited by the serine/threonine kinase inhibitor, H7.28Thrombin-stimulated platelet activation and aggregation requires the activation of a MAPK kinase and the phosphorylation of a serine/threonine kinase.30 Thus, phosphorylated MAPK kinases and serine/threonine kinases are required for the up-regulation of Ang-2 in HUVE cells, whereas the PI-3 kinase pathway does not appear to be involved.

It is generally accepted that VEGF, Ang-1, and Ang-2 are necessary for efficient blood vessel growth and development. It has been proposed that Ang-2, a natural antagonist of Ang-1, may be an important proangiogenic factor in that it may counteract Ang-1–mediated blood vessel stability, thus maintaining the endothelium in a more plastic state and promoting the response of endothelial cells to angiogenesis growth factors.50 Up-regulation of both VEGF and Ang-2 by thrombin indicates that angiogenesis might be facilitated by thrombosis. Thus, the well-described association of thrombosis with cancer16-20 may be contributing to tumorigenesis by the initiation of thrombin-stimulated angiogenesis, which could explain, at least in part, the enhancement of experimental tumorigenesis by thrombin.21-25 

Supported by NIH grant HL-13336-28, the Hildegarde D. Becher Foundation, and grants from the Helen Polonsky Research Fund, Dorothy and Seymour Weinstein Research Fund, and New York Community Trust.

The publication costs of this article were defrayed in part by page charge payment. Therefore, and solely to indicate this fact, this article is hereby marked “advertisement” in accordance with 18 U.S.C. section 1734.

1
Folkman
J
Angiogenesis in cancer, vascular, rheumatoid and other disease.
Nat Med.
1
1995
27
31
2
Folkman
J
Shing
Y
Angiogenesis.
J Biol Chem.
267
1992
10931
10934
3
Pepper
MS
Ferrara
N
Orci
L
Montesano
R
Potent synergism between vascular endothelial growth factor and basic fibroblast growth factor in the induction of angiogenesis in vitro.
Biochem Biophys Res Commun.
189
1992
824
831
4
Eriksson
U
Alitalo
K
Structure, expression and receptor-binding properties of novel vascular endothelial growth factors.
Curr Top Microbiol Immunol.
237
1999
41
57
5
Veikkola
T
Karkainen
MJ
Claesson-Welsh
L
Alitalo
K
Regulation of angiogenesis via vascular endothelial growth factor receptors.
Cancer Res.
60
2000
203
212
6
Davis
S
Aldrich
TH
Jones
PF
et al
Isolation of angiopoietin-1, a ligand for the TIE-2 receptor, by secretion-trap expression cloning.
Cell.
87
1996
1161
1169
7
Suri
C
Jones
PF
Patan
S
et al
Requisite role of angiopoietin-1, a ligand for the TIE-2 receptor, during embryonic angiogenesis.
Cell.
87
1996
1171
1180
8
Thurston
G
Suri
C
Smith
K
et al
Leakage-resistant blood vessels in mice transgenically overexpressing angiopoietin-1.
Science.
286
1999
2511
2514
9
Maisonpierre
PC
Suri
C
Jones
PF
et al
Angiopoietin-2, a natural antagonist for Tie-2 that disrupts in vivo angiogenesis.
Science.
277
1997
55
60
10
Koblizek
TI
Weiss
C
Yancopoulos
GD
Deutsch
U
Risau
W
Angiopoietin-1 induces sprouting angiogenesis in vitro.
Curr Biol.
8
1998
529
532
11
Oh
H
Takagi
H
Suzuma
K
Otani
A
Matsumura
M
Honda
Y
Hypoxia and vascular endothelial growth factor selectively up-regulate angiopoietin-2 in bovine microvascular endothelial cells.
J Biol Chem.
274
1999
15732
15739
12
Haralabopoulos
GC
Grant
DS
Kleinman
HK
Maragoudakis
ME
Thrombin promotes endothelial cell alignment in matrigel in vitro and angiogenesis in vivo.
Am J Physiol.
273
1997
239
245
13
Herbert
JM
Dupuy
E
Laplace
MC
Zini
JM
Bar Shavit
R
Thrombin induces endothelial cell growth via both a proteolytic and a non-proteolytic pathway.
Biochem J.
303
1994
227
231
14
Tsopanoglou
NE
Maragoudakis
ME
On the mechanism of thrombin-induced angiogenesis. Potentiation of vascular endothelial growth factor activity on endothelial cells by up-regulation of its receptors.
J Biol Chem.
274
1999
23969
23976
15
Tsopanoglou
NE
Pipili-Synetos
E
Maragoudakis
ME
Thrombin promotes angiogenesis by a mechanism independent of fibrin formation.
Am J Physiol.
264
1993
C1302
C1307
16
Trousseau
A
Phelgmasia alba dolens. Clinique Medicale de Hotel-Dieu de Paris
3
1865
94
New Syndenham Society.
London
17
Merskey
C
Johnson
AJ
Harris
JU
Want
MT
Swain
S
Isolation of fibrinogen-fibrin related antigen from human plasma by immune-affinity chromatography: its characterization in normal subjects and in defibrinating patients with abruptio placentae and disseminated cancer.
Br J Haematol.
44
1980
655
670
18
Peuscher
FW
Cleton
FJ
Armstrong
L
Stoepman van Dalen
EA
van Aken
WG
Significance of plasma fibrinopeptide A (FPA) in patients with malignancy.
J Lab Clin Med.
96
1980
5
14
19
Rickles
FR
Edwards
RC
Barb
C
Cronlund
M
Abnormalities of blood coagulation in patients with cancer: fibrinopeptide A generation and tumor growth.
Cancer.
51
1983
301
307
20
Yoda
Y
Abe
T
Fibrinopeptide A (FPA) level and fibrinogen kinetics in patients with malignant disease.
Thromb Haemost.
46
1981
706
709
21
Klepfish
A
Greco
MA
Karpatkin
S
Thrombin stimulates melanoma tumor cell binding to endothelial cells and subendothelial matrix.
Int J Cancer.
53
1993
978
982
22
Nierodzik
ML
Bain
RM
Liu
L-X
Shivji
M
Takeshita
K
Karpatkin
S
Presence of the seven transmembrane thrombin receptor on human tumour cells: effect of activation on tumour adhesion to platelets and tumour tyrosine phosphorylation.
Br J Haematol.
92
1996
452
457
23
Nierodzik
ML
Chen
K
Takeshita
K
et al
Protease-activated receptor 1 (PAR-1) is required and rate-limiting for thrombin-enhanced experimental pulmonary metastasis.
Blood.
92
1998
3694
3700
24
Nierodzik
ML
Kajumo
F
Karpatkin
S
Effect of thrombin treatment of tumor cells on adhesion of tumor cells to platelets in vitro and metastasis in vivo.
Cancer Res.
52
1992
3267
3272
25
Nierodzik
ML
Plotkin
A
Kajumo
F
Karpatkin
S
Thrombin stimulates tumor-platelet adhesion in vitro and metastasis in vivo.
J Clin Invest.
87
1991
229
236
26
Grand
RJA
Turnell
AS
Grabham
PW
Cellular consequences of thrombin-receptor activation.
Biochem J.
313
1996
353
368
27
Huang
Y-Q
Li
J-J
Karpatkin
S
Identification of a family of alternatively spliced mRNA species of angiopoietin-1.
Blood.
95
2000
1993
1999
28
Donovan
FM
Cunningham
DD
Signaling pathways involved in thrombin-induced cell protection.
J Biol Chem.
273
1998
12746
12752
29
Gutkind
JS
Lacal
PM
Robbins
KC
Thrombin-dependent association of phosphatidylinositol-3 kinase with p60c-src and p59fyn in human platelets.
Mol Cell Biol.
10
1990
3806
3809
30
Papkoff
J
Chen
RH
Blenis
J
Forsman
J
p42 mitogen-activated protein kinase and p90 ribosomal S6 kinase are selectively phosphorylated and activated during thrombin-induced platelet activation and aggregation.
Mol Cell Biol.
14
1994
463
472
31
Mohle
R
Green
D
Moore
MAS
Nachman
RL
Raffi
S
Constitutive production and thrombin-induced release of VEGF by human megakaryocytes and platelets.
Proc Natl Acad Sci U S A.
94
1997
663
668
32
Moloney
JP
Sillimon
CC
Ambruso
DR
Wang
J
Tuder
RM
Voelkel
NF
In vitro release of VEGF during platelet aggregation.
Am J Phys.
275
1988
H1054
33
Li
J-J
Huang
Y-Q
Basch
R
Karpatkin
S
Thrombin induces the release of angiopoietin-1 from platelets.
Thromb Haemost.
85
2001
204
206
34
Benjamin
LE
Keshet
E
Conditional switching of vascular endothelial growth factor (VEGF) expression in tumors: induction of endothelial cell shedding and regression of hemangioblastoma-like vessels by VEGF withdrawal.
Proc Natl Acad Sci U S A.
94
1997
8761
8766
35
Plate
KH
Breier
G
Weich
HA
Risau
W
Vascular endothelial growth factor is a potential tumour angiogenesis factor in human gliomas in vivo.
Nature.
359
1992
845
848
36
Shweiki
D
Itin
A
Soffer
D
Keshet
E
Vascular endothelial growth factor induced by hypoxia may mediate hypoxia-initiated angiogenesis.
Nature.
359
1992
843
845
37
Zacharski
LR
Memoli
VA
Morain
WD
Schlaeppi
J-M
Rousseau
SM
Cellular localization of enzymatically active thrombin in intact tissue by hirudin binding.
Thromb Haemost.
73
1995
793
797
38
Kim
KJ
Li
B
Winer
J
Armanini
M
et al
Inhibition of vascular endothelial growth factor induced angiogenesis suppresses tumour growth in vivo.
Nature.
362
1993
841
844
39
Kondo
S
Asano
M
Suzuki
H
Significance of vascular endothelial growth factor/vascular permeability factor for solid tumor growth, and its inhibition by the antibody.
Biochem Biophys Res Commun.
194
1993
1234
1241
40
Lin
P
Buxton
JA
Acheson
A
et al
Antiangiogenic gene therapy targeting the endothelium-specific receptor tyrosine kinse Tie-2.
Proc Natl Acad Sci U S A.
95
1998
8829
8834
41
Tanaka
S
Mori
M
Sakamoto
Y
Makuuchi
M
Sugimachi
K
Wands
JR
Biologic significance of angiopoietin-2 expression in human hepatocellular carcinoma.
J Clin Invest.
103
1999
341
345
42
Eggert
A
Ikegaki
N
Kwiatkowski
J
Zhao
H
Brodeur
GM
Himelstein
BP
High-level expression of angiogenic factors is associated with advanced tumor stage in human neuroblastomas.
Clin Cancer Res.
6
2000
1900
1908
43
Wong
MP
Chan
SY
Fu
KH
et al
The angiopoietins, Tie2 and vascular endothelial growth factor are differentially expressed in the transformation of normal lung to non-small cell lung carcinomas.
Lung Cancer.
29
2000
11
22
44
Ijland
SA
Jager
MJ
Heijdra
BM
Westphal
JR
Peek
R
Expression of angiogenic and immunosuppressive factors by uveal melanoma cell lines.
Melanoma Res.
9
1999
445
450
45
Bunone
G
Vigneri
P
Mariani
L
et al
Expression of angiogenesis stimulators and inhibitors in human thyroid tumors and correlation with clinical pathological features.
Am J Pathol.
155
1999
1967
1976
46
Stratmann
A
Risau
W
Plate
KH
Cell type-specific expression of angiopoietin-1 and angiopoietin-2 suggests a role in glioblastoma angiogenesis.
Am J Pathol.
153
1998
1333
1339
47
Huang
Y-Q
Li
J-J
Hu
L
Karpatkin
S
Thrombin induces vascular endothelial cell growth factor (VEGF) in human tumor cells and fibroblasts [abstract].
Blood.
94(suppl 1)
1999
12a
48
Huang
Y-Q
Li
J-J
Hu
L
Lee
M
Karpatkin
S
Thrombin induces increased expression and secretion of VEGF from human FS4 fibroblasts, DU145 prostate cells and CHRF megakaryocytes.
Thromb Haemost.
86
2001
1094
1098
49
Ollivier
V
Chabbat
J
Herbert
JM
Hakim
J
de Prost
D
Vascular endothelial growth factor production by fibroblasts in response to factor VIIa binding to tissue factor involves thrombin and factor Xa.
Arterioscler Thromb Vasc Biol.
20
2000
1374
1381
50
Holash
J
Maisonpierre
PC
Compton
D
et al
Vessel cooption, regression, and growth in tumors mediated by angiopoietins and VEGF.
Science.
284
1999
1994
1998

Author notes

Yao-Qi Huang or Simon Karpatkin, Dept of Medicine and Kaplan Cancer Center, New York University Medical School, 550 First Ave, New York, NY 10016; e-mail: huangy02@med.nyu.edu orsimon.karpatkin@med.nyu.edu.

Sign in via your Institution